Roux-en-Y gastric bypass (RYGB) has been shown to inhibit β-cell apoptosis, but the underlying mechanisms are not yet fully understood. Cytochrome c oxidase subunit 6A2 (COX6A2) is expressed in β-cells. Here, we investigated the role of COX6A2 in β-cell apoptosis, especially following RYGB. We found that RYGB significantly reduced β-cell apoptosis, accompanied by decreased COX6A2 expression in islets from diabetic Goto-Kakizaki (GK) rats. It is noteworthy that overexpression of COX6A2 promoted β-cell apoptosis, whereas COX6A2 deficiency suppressed it, suggesting the proapoptotic role of COX6A2 in β-cells. Mechanistically, increased COX6A2 interacted with and upregulated the expression of cyclophilin D (CypD), facilitating the release of cytochrome c from mitochondria to the cytoplasm, thereby promoting β-cell apoptosis. Furthermore, high glucose–activated carbohydrate-responsive element-binding protein (ChREBP) epigenetically regulated COX6A2 expression by recruiting histone acetyltransferase p300 to augment histone H3 acetylation at the Cox6a2 promoter, a process inhibited by glucagon-like peptide 1 (GLP-1) signaling. Given that RYGB enhances GLP-1 signaling, RYGB is likely to deactivate ChREBP by boosting GLP-1/cAMP-dependent protein kinase (PKA) signaling, thereby reducing COX6A2 expression in islets from GK rats. These findings highlight the crucial role of the GLP-1/PKA/ChREBP axis-controlled COX6A2 in β-cell apoptosis, revealing a previously unrecognized mechanism underlying the reduction in β-cell apoptosis induced by RYGB.

Article Highlights

  • Cytochrome c oxidase subunit 6A2 (COX6A2) expression is increased in diabetic islets.

  • Increased COX6A2 promotes β-cell apoptosis via modulation of cyclophilin D-mediated cytochrome c release from mitochondria to the cytoplasm.

  • Carbohydrate-responsive element-binding protein epigenetically regulates COX6A2 expression in β-cells.

  • Roux-en-Y gastric bypass reduces COX6A2 expression by regulating the glucagon-like peptide 1/cAMP-dependent protein kinase/carbohydrate-responsive element-binding protein signaling pathway.

Pancreatic β-cell apoptosis plays a key role in the development of type 2 diabetes (T2D). Bariatric surgery, such as Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy, has been demonstrated to effectively treat T2D and improve β-cell function (1–3). RYGB has been shown to suppress β-cell apoptosis (4,5), which contributes to the increased β-cell mass and subsequent remission of T2D (3). However, the mechanisms underlying improved β-cell apoptosis after RYGB remain unclear.

Glucagon-like peptide 1 (GLP-1) is an incretin released from enteroendocrine L cells. It amplifies the glucose-stimulated insulin secretion (GSIS) of β-cells via activation of intracellular cAMP/cAMP-dependent protein kinase (PKA) and cAMP/Epac2 signaling pathway (6). GLP-1 has also been reported to protect β-cells from apoptosis under diabetic conditions (7). We and others have demonstrated that RYGB increases the plasma GLP-1 level (8) and islet GLP-1R expression (9,10), suggesting that RYGB enhances the GLP-1 signaling pathway. However, whether the enhanced GLP-1 signaling is involved in RYGB-decreased β-cell apoptosis is largely unknown.

Carbohydrate response element binding protein (ChREBP, also known as Mlxipl), a glucose-sensing transcription factor, is expressed in multiple tissues, including pancreatic islets (11). ChREBP comprises two isoforms, ChREBPα and ChREBPβ (hereafter, ChREBP refers to ChREBPα). Upon exposure to high glucose, ChREBP is activated and mediates glucose-induced gene expression and β-cell apoptosis (12). After activation, ChREBP translocates to the nucleus and binds to the carbohydrate response element (ChoRE) on the target gene promoter, thereby recruiting coactivator, such as histone acetyltransferase p300, leading to enhanced histone acetylation at the target gene promoter and culminating in elevated target gene transcription (13). The transcriptional activity of ChREBP is contingent on its nuclear localization controlled by the phosphorylation of specific amino acid residues, such as Ser196, Ser568, and Thr666, which are phosphorylated by PKA and AMPK (14).

Cytochrome c oxidase subunit 6A2 (COX6A2) is expressed in pancreatic β-cells (15). Previous studies have shown that COX6A2 regulates cell apoptosis-related reactive oxygen species generation in skeletal muscles (16) and neurons (17). Deficiency of Cox6a2 protects mice against high-fat diet–induced obesity and insulin resistance (16). Currently, the role of COX6A2 in β-cell apoptosis, especially whether it is involved in RYGB-decreased β-cell apoptosis, is still elusive. Here we show that β-cell apoptosis and COX6A2 expression are increased in diabetic Goto-Kakizaki (GK) rat islets, whereas both are inhibited following RYGB surgery. Increased COX6A2 promotes β-cell apoptosis by modulating cyclophilin D (CypD)-mediated release of cytochrome c from mitochondria. Additionally, COX6A2 expression is epigenetically controlled by ChREBP. RYGB enhances GLP-1 signaling to phosphorylate and deactivate ChREBP, thereby inhibiting COX6A2 expression and subsequently alleviating β-cell apoptosis.

Experimental Animals and Isolation of Islets

Male Wistar and GK rats (aged 8 weeks) were procured from SLRC (Shanghai, China), and 6-week-old male Sprague Dawley (SD) rats came from the Guangdong Medical Laboratory Animal Center. The GK and SD rats underwent Roux-en-Y gastric bypass (RYGB) or sham surgery. Additionally, some GK rats were given intraperitoneal injections of exendin-4 (10 μg/kg body wt/day) (18) or saline for 8 days. Cox6a2 knockout (KO) mice (C57BL/6JGpt) were generated using clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) technology to delete exons 1 to 3 in the Cox6a2 gene and were obtained from GemPharmatech Company (Nanjing, China). Because estrogen regulates the expression of COX6A2 (19), to avoid the interference of estrogen, male wild-type (WT) and Cox6a2 KO mice (aged 5 weeks) were used in this study. These mice were fed a high-fat diet for 3 months, followed by intraperitoneal injection of streptozotocin (STZ) (30 mg/kg body wt/day) (20) for 3 consecutive days to induce a diabetic state. The animal procedures followed the principles of laboratory animal care and were approved by the Shenzhen University Animal Care Committee. The pancreata of the rats and mice were processed by sectioning or by enzymatic digestion using collagenase P to isolate islets for examination.

In Vivo Administration of Adeno-Associated Virus

The adeno-associated virus (AAV) AAV9-Cox6a2-KO plasmid was generated by inserting the single guide (sg)RNA (5′-TAAGGTCCTCAGTCGGAGCA-3′) target against the rat Cox6a2 gene into pX601-AAV-CMV-SaCas9-3xHA-U6-sgRNA vector. The AAV was packaged and provided by Vigene Biosciences (Jinan, China). To decrease COX6A2 expression in islets, GK rats were anesthetized with inhalation of 2–3% isoflurane and injected with 50 μL of AAV containing ∼2 × 1012 Cox6a2-KO virus particles via the common bile duct. The control rats were injected with 50 μL of AAV control virus particles containing a nonspecific sgRNA. All rats were sacrificed 2 weeks later for biochemical analysis.

RYGB and Sham Surgery

The RYGB and sham procedures were performed as previously described (21). Briefly, the rats were anesthetized with isoflurane after an overnight fast. The jejunum was detached at a distance of 16 cm from the Treitz ligament. The proximal segment was anastomosed to the 30 cm of the Roux limb, and the distal portion was stitched to the residual stomach. Some GK-RYGB rats received daily intraperitoneal injections of exendin(9-39) (30 μg/kg body wt) (22) or saline for 10 days.

Intravenous Glucose Tolerance Test

The intravenous glucose tolerance test was performed as described previously (21). In brief, after an intravenous injection of 1.5 g/kg glucose, blood samples were taken from the jugular veins of rats to determine plasma insulin levels.

Intraperitoneal Glucose Tolerance Test

After overnight fasting, the mice were intraperitoneally injected with glucose (2 g/kg). Blood glucose levels were measured at 0, 15, 30, 60, 90, and 120 min after glucose administration.

Cell Lines

INS-1 832/13 cells were cultured in RPMI 1640 medium (23). Rat Cox6a2 or CypD coding sequences were amplified by PCR using the primers listed in Supplementary Table 1, and the PCR products were inserted into the retroviral pMX-puro vector. INS-1 832/13 cells were transduced with pMX-puro-vector, pMX-puro-COX6A2, or pMX-puro-CypD, followed by selection with 3 μg/mL puromycin for 1 week. For the knockdown of ChREBP or COX6A2, INS-1 832/13 cells were transduced with either scramble or shChREBP (cat. RMM3981-201916728) or shCOX6A2 (cat. RMM3981-201787879) plasmid from GE Dharmacon.

Western Blotting Analysis

The experiments were performed as previously described (23). The following antibodies were used: phospho-ChREBP (Ser196) (Proteintech), ChREBP (NB400-135, Novus), COX6A2 (11421-1-AP, Proteintech), cytochrome c (no. 11940, Cell Signaling Technology), COX IV (no. 4850, Cell Signaling Technology), cleaved caspase-3 (no. 9661, Cell Signaling Technology), caspase-9 (no. 9508, Cell Signaling Technology), CypD (ab110324, Abcam), GAPDH (no. 5174, Cell Signaling Technology), histone H3 (ab1791, Abcam), and β-actin (A5441, Sigma-Aldrich). The densities of the immunoblot bands were determined by Gel-Pro Analyzer 4.0 software.

RNA Extraction and Real-Time PCR

RNA was extracted from INS-1 832/13 cells and islets using the Trizol method. Quantitative PCR analyses were conducted using the SYBR Green master mix from Promega on the ABI QuantStudio 5 system. Primer sequences used can be found in Supplementary Table 2. The quantity of target gene mRNA was normalized to GAPDH or β-actin.

Construction of Luciferase Plasmid and Promoter-Reporter Assay

Rat promoter fragments of Cox6a2 (−185 base pair [bp] to +65 bp) were amplified by PCR using primers shown in Supplementary Table 1 and subsequently cloned into a pGL3-basic plasmid (Promega). Site-directed mutagenesis of the putative ChREBP binding site from −20 bp to −15 bp in the Cox6a2 promoter was accomplished using the QuikChange II Mutagenesis Kit (Agilent Technology) with the primers listed in Supplementary Table 1. For determination of Cox6a2 promoter activity, human embryonic kidney 293 T cells were cotransfected with pGL3-Cox6a2 plasmid and pcDNA3.1 or ChREBP-overexpressing plasmid (no. 39235, Addgene), along with Renilla luciferase plasmid. After 48 h, the resulting promoter activity was measured using the Dual-Luciferase Reporter Assay kit (Promega).

Chromatin Immunoprecipitation Assay

Chromatin immunoprecipitation (ChIP) assays were performed as previously reported (21). Chromatin was immunoprecipitated with antibodies (3 μg) against ChREBP (NB400-135, Novus), acetylated histone H3 (06-599, Sigma-Aldrich) and p300 (ab14984, Abcam), respectively. The obtained DNA fragments were quantified by qPCR using the primers listed in Supplementary Table 3.

Coimmunoprecipitation

INS-1 832/13 cells were lysed in immunoprecipitation lysis buffer containing protease inhibitors. Cell lysates (1 mg) were incubated with 3 μg of CypD antibody overnight at 4°C. The complexes were immunoprecipitated with 60 μL protein A agarose beads for 3 h at 4°C and then washed sequentially with low-salt wash buffer, high-salt wash buffer, LiCl wash buffer, and Tris-HCl/EDTA buffer. The immunoprecipitated supernatants were separated by SDS-PAGE, followed by immunoblotting with COX6A2 antibody, and detected using the Tanon 5200 image system.

Immunofluorescence

To determine the colocalization of COX6A2 and CypD, INS-1 832/13 cells were fixed with paraformaldehyde and permeabilized with Triton X-100. The cells were then incubated overnight at 4°C with primary antibodies against COX6A2 and CypD, followed by incubation with fluorescent secondary antibodies. Images were captured using a Zeiss LSM 510 confocal laser scanning microscope.

Additionally, pancreatic sections were stained with cleaved caspase-3, insulin, and DAPI. The resulting fluorescence was visualized using the Zeiss Axio Observer 3 fluorescent microscope. The insulin-positive and DAPI-stained areas were calculated using ImageJ software. β-Cell area was expressed as the ratio of the insulin-positive area to the DAPI-stained area. β-Cell mass was calculated through the β-cell area multiplied by the corresponding pancreatic weight.

Mitochondrial Transmembrane Potential

The mitochondrial transmembrane potential (ΔΨm) of INS-1 832/13 cells was analyzed by JC-1 Mitochondrial Membrane Potential Assay Kit (Beyotime Biotechnology, Shanghai, China). The cells were incubated with JC-1 (1:200) dye for 20 min at 37°C. The cell-associated fluorescence was measured with an Axio Observer 3 microscope.

TUNEL Assays

TUNEL assays were performed using a TUNEL assay kit (Beyotime Biotechnology). The samples were counterstained with DAPI to visualize the cell nucleus. Additionally, pancreatic sections were immunoassayed with anti-insulin antibodies to identify β-cells. The samples were observed using an Axio Observer 3 microscope. TUNEL-positive cells were counted and normalized to total β-cells.

Flow Cytometry

INS-1 832/13 cells were collected after digestion with 0.25% trypsin. The cells were incubated with a mixture of propidium iodide and annexin V (Beyotime, Shanghai, China) for 20 min at room temperature, followed by immediate examination using flow cytometry.

Adenovirus-Mediated Cox6a2 Gene Transfer to Islets

Isolated islets were prepared for infection by adding 8 μg/mL polybrene. The islets were infected with adenovirus (Ad)-green fluorescent protein (GFP)-COX6A2 or Ad-GFP adenovirus (Sangon Biotech, Shanghai, China) at a multiplicity of infection of 500 for 8 h. Then the islets were transferred to a fresh medium. After 72 h of culture, the islets were harvested for Western blotting analysis.

Statistical Analyses

The statistical analysis was performed using SPSS 20.0 software. The independent t test was used for comparing two groups, and one-way ANOVA with the least significant difference post hoc test was used for comparing more than two groups. A significance level of P < 0.05 was considered statistically significant.

Data and Resource Availability

The data sets generated during the current study are available upon request. The resources are available upon request.

RYGB Inhibits β-Cell Apoptosis and COX6A2 Expression in GK Rats

The level of cleaved caspase-3 was examined 1 month after surgery to monitor islet cell apoptosis. As shown in Fig. 1A, RYGB markedly decreased apoptosis in islet cell populations in GK rats, consistent with previous reports (4,24). Correspondently, RYGB increased insulin-positive cell area (Fig. 1B and C), improved GSIS (Fig. 1D), and led to the remission of hyperglycemia in GK rats (Fig. 1E). We next determined the effect of RYGB on COX6A2 expression. Compared with Wistar islets, GK rat islets displayed higher COX6A2 mRNA and protein levels, respectively (Fig. 1F and G). Notably, the increased expression of COX6A2 was inhibited by RYGB (Fig. 1F and G). Comparable results were also obtained in SD-RYGB rats (Fig. 1H and I), indicating that RYGB could suppress COX6A2 expression in rat islets.

Figure 1

RYGB decreases COX6A2 expression in islets from GK and SD rats. A: The level of cleaved caspase-3 in islets from Wistar, GK-sham, and GK-RYGB rats. β-Actin was used as a loading control. Data are mean ± SEM (n = 6). *P < 0.05, **P < 0.01. B: Immunofluorescence of insulin and DAPI in pancreata of Wistar, GK-sham, and GK-RYGB rats. Scale bar, 200 μm. C: Quantification of insulin-positive cell area in B. Data are mean ± SEM (n = 6 rats per group). *P < 0.05, **P < 0.01. D: Plasma insulin in Wistar, GK-sham, and GK-RYGB rats. Data are mean ± SEM (n = 6–8 rats per group). *P < 0.05 vs. GK-sham. E: Plasma glucose in Wistar, GK-sham, and GK-RYGB rats. Data are mean ± SEM (n = 6). *P < 0.05, **P < 0.01 vs. GK-sham. The level of COX6A2 mRNA (F) and protein (G) in islets from Wistar, GK-sham, and GK-RYGB rats. β-Actin was used as a loading control. Data are mean ± SEM (n = 6–7). *P < 0.05, **P < 0.01. The level of COX6A2 mRNA (H) and protein (I) in islets from SD-sham and SD-RYGB rats. GAPDH was used as a loading control. Data are mean ± SEM (n = 6). **P < 0.01.

Figure 1

RYGB decreases COX6A2 expression in islets from GK and SD rats. A: The level of cleaved caspase-3 in islets from Wistar, GK-sham, and GK-RYGB rats. β-Actin was used as a loading control. Data are mean ± SEM (n = 6). *P < 0.05, **P < 0.01. B: Immunofluorescence of insulin and DAPI in pancreata of Wistar, GK-sham, and GK-RYGB rats. Scale bar, 200 μm. C: Quantification of insulin-positive cell area in B. Data are mean ± SEM (n = 6 rats per group). *P < 0.05, **P < 0.01. D: Plasma insulin in Wistar, GK-sham, and GK-RYGB rats. Data are mean ± SEM (n = 6–8 rats per group). *P < 0.05 vs. GK-sham. E: Plasma glucose in Wistar, GK-sham, and GK-RYGB rats. Data are mean ± SEM (n = 6). *P < 0.05, **P < 0.01 vs. GK-sham. The level of COX6A2 mRNA (F) and protein (G) in islets from Wistar, GK-sham, and GK-RYGB rats. β-Actin was used as a loading control. Data are mean ± SEM (n = 6–7). *P < 0.05, **P < 0.01. The level of COX6A2 mRNA (H) and protein (I) in islets from SD-sham and SD-RYGB rats. GAPDH was used as a loading control. Data are mean ± SEM (n = 6). **P < 0.01.

Close modal

Increased COX6A2 Facilitates β-Cell Apoptosis

To investigate the role of increased COX6A2 in β-cell apoptosis, mouse islets were infected with Ad-GFP or Ad-GFP-COX6A2 to enhance COX6A2 expression. The infection with Ad-GFP-COX6A2 resulted in increased COX6A2 expression (Supplementary Fig. 1A) and an elevated level of cleaved caspase-3 (Fig. 2A). Similar results were also made in COX6A2-overexpressing INS-1 832/13 cells (Fig. 2Band Supplementary Fig. 1B). The increased apoptosis in COX6A2-overexpressing cells was also confirmed by TUNEL staining (Fig. 2C and D) and flow cytometry (Fig. 2E and F), respectively.

Figure 2

COX6A2 regulates β-cell apoptosis. A: Western blotting analysis of cleaved caspase-3 levels in islets infected with Ad-GFP or Ad-GFP-COX6A2 for 72 h. GAPDH was used as a loading control. Data are mean ± SEM (n = 6). *P < 0.05. B: The protein level of cleaved caspase-3 in vector and COX6A2-overexpressing INS-1 832/13 cells. β-Actin was used as a loading control. Data are mean ± SEM (n = 6). **P < 0.01. C: Cell apoptosis assessed by TUNEL staining in vector and COX6A2-overexpressing INS-1 832/13 cells. Arrowheads indicate apoptotic cells. Scale bar, 50 μm. D: Quantification of the TUNEL-positive cell rate in C. Data are mean ± SEM (n = 7 independent experiments). **P < 0.01. E: The vector and COX6A2-overexpressing INS-1 832/13 cells were stained with propidium iodide (PI) and annexin V, followed by flow cytometry analysis. F: Statistical analysis of apoptotic cells in E. Data are mean ± SEM (n = 6 independent experiments). **P < 0.01. G: The level of cleaved caspase-3 in the scramble and COX6A2 knockdown INS-1 832/13 cells cultured in G5.5 or G30 for 4 days. β-Actin was used as a loading control. Data are mean ± SEM (n = 6). **P < 0.01. H: The scramble and COX6A2 knockdown INS-1 832/13 cells were cultured in G5.5 or G30 for 4 days, followed by determination of cell apoptosis with TUNEL assay. Arrowheads indicate TUNEL-positive cells. Scale bar, 20 μm. I: Quantification of the apoptotic cell rate in H. Data are mean ± SEM (n = 6 independent experiments). **P < 0.01.

Figure 2

COX6A2 regulates β-cell apoptosis. A: Western blotting analysis of cleaved caspase-3 levels in islets infected with Ad-GFP or Ad-GFP-COX6A2 for 72 h. GAPDH was used as a loading control. Data are mean ± SEM (n = 6). *P < 0.05. B: The protein level of cleaved caspase-3 in vector and COX6A2-overexpressing INS-1 832/13 cells. β-Actin was used as a loading control. Data are mean ± SEM (n = 6). **P < 0.01. C: Cell apoptosis assessed by TUNEL staining in vector and COX6A2-overexpressing INS-1 832/13 cells. Arrowheads indicate apoptotic cells. Scale bar, 50 μm. D: Quantification of the TUNEL-positive cell rate in C. Data are mean ± SEM (n = 7 independent experiments). **P < 0.01. E: The vector and COX6A2-overexpressing INS-1 832/13 cells were stained with propidium iodide (PI) and annexin V, followed by flow cytometry analysis. F: Statistical analysis of apoptotic cells in E. Data are mean ± SEM (n = 6 independent experiments). **P < 0.01. G: The level of cleaved caspase-3 in the scramble and COX6A2 knockdown INS-1 832/13 cells cultured in G5.5 or G30 for 4 days. β-Actin was used as a loading control. Data are mean ± SEM (n = 6). **P < 0.01. H: The scramble and COX6A2 knockdown INS-1 832/13 cells were cultured in G5.5 or G30 for 4 days, followed by determination of cell apoptosis with TUNEL assay. Arrowheads indicate TUNEL-positive cells. Scale bar, 20 μm. I: Quantification of the apoptotic cell rate in H. Data are mean ± SEM (n = 6 independent experiments). **P < 0.01.

Close modal

To further explore the role of COX6A2 in cell apoptosis, we also generated the scramble and COX6A2 knockdown (shCOX6A2) INS-1 832/13 cells (Supplementary Fig. 1C) and cultured these cells in 5.5 mmol/L glucose (G5.5) or 30 mmol/L glucose (G30) medium for 4 days. Consistent with the previous study (25), G30 culture enhanced cleaved caspase-3 levels in scramble cells (Fig. 2G). Of note, shCOX6A2 markedly reduced the increase of cleaved caspase-3 induced by high glucose (Fig. 2G). Thus, apoptosis caused by G30, as determined by the TUNEL assay, was blocked by shCOX6A2 (Fig. 2H and I). Similarly, COX6A2 deficiency diminished the STZ-induced cell apoptosis (Supplementary Fig. 2A and B).

To investigate the effect of COX6A2 on diabetic β-cell apoptosis in vivo, the WT and Cox6a2 KO mice were fed a high-fat diet and treated with STZ. The absence of COX6A2 expression in islets from Cox6a2 KO mice confirmed the knockout, as opposed to its detection in WT mice (Fig. 3A). Intriguingly, the level of cleaved caspase-3 was reduced in the KO islets compared with the WT islets (Fig. 3A and B). These data suggest that increased COX6A2 accelerates diabetic islet cell apoptosis. In line with the decreased apoptosis in Cox6a2 KO islets, the insulin-positive cell area was increased in KO mice (Fig. 3C and D). Consequently, Cox6a2 KO mice exhibited enhanced insulin levels (Fig. 3E and F), improved glycemic control (Fig. 3G and H), and reduced blood glucose levels (Fig. 3I).

Figure 3

Cox6a2 KO reduces β-cell apoptosis induced by a high-fat diet and STZ. A–I: After 3 months of being fed a high-fat diet, Cox6a2+/+ and Cox6a2−/− mice were intraperitoneally injected with 30 mg/kg body wt STZ for 3 consecutive days. A: The protein expression of COX6A2 and cleaved caspase-3 were analyzed in islets from Cox6a2+/+ and Cox6a2−/− mice. GAPDH and β-actin were used as a loading control. B: Statistical analysis of the protein levels in A. Data are mean ± SEM (n = 7 mice per group). **P < 0.01. C: Immunofluorescence of insulin and DAPI in pancreata of Cox6a2+/+ and Cox6a2−/− mice. Scale bar, 200 μm. D: Quantification of insulin-positive cell area in C. Data are mean ± SEM (n = 6 mice per group). *P < 0.05. E: Plasma insulin levels were measured in Cox6a2+/+ and Cox6a2−/− mice following an intraperitoneal injection of glucose (2 g/kg body wt). Data are mean ± SEM (n = 6 mice per group). **P < 0.01. F: The areas under curve (AUC) for insulin response from E were calculated. Data are mean ± SEM (n = 6). **P < 0.01. G: Plasma glucose levels were measured during intraperitoneal glucose tolerance test in Cox6a2+/+ and Cox6a2−/− mice. Data are mean ± SEM (n = 6). *P < 0.05; **P < 0.01. H: AUC calculations for glucose in G. Data are mean ± SEM (n = 6). **P < 0.01. I: Blood glucose in Cox6a2+/+ and Cox6a2−/− mice after STZ injection. Data are mean ± SEM (n = 6). *P < 0.05; **P < 0.01.

Figure 3

Cox6a2 KO reduces β-cell apoptosis induced by a high-fat diet and STZ. A–I: After 3 months of being fed a high-fat diet, Cox6a2+/+ and Cox6a2−/− mice were intraperitoneally injected with 30 mg/kg body wt STZ for 3 consecutive days. A: The protein expression of COX6A2 and cleaved caspase-3 were analyzed in islets from Cox6a2+/+ and Cox6a2−/− mice. GAPDH and β-actin were used as a loading control. B: Statistical analysis of the protein levels in A. Data are mean ± SEM (n = 7 mice per group). **P < 0.01. C: Immunofluorescence of insulin and DAPI in pancreata of Cox6a2+/+ and Cox6a2−/− mice. Scale bar, 200 μm. D: Quantification of insulin-positive cell area in C. Data are mean ± SEM (n = 6 mice per group). *P < 0.05. E: Plasma insulin levels were measured in Cox6a2+/+ and Cox6a2−/− mice following an intraperitoneal injection of glucose (2 g/kg body wt). Data are mean ± SEM (n = 6 mice per group). **P < 0.01. F: The areas under curve (AUC) for insulin response from E were calculated. Data are mean ± SEM (n = 6). **P < 0.01. G: Plasma glucose levels were measured during intraperitoneal glucose tolerance test in Cox6a2+/+ and Cox6a2−/− mice. Data are mean ± SEM (n = 6). *P < 0.05; **P < 0.01. H: AUC calculations for glucose in G. Data are mean ± SEM (n = 6). **P < 0.01. I: Blood glucose in Cox6a2+/+ and Cox6a2−/− mice after STZ injection. Data are mean ± SEM (n = 6). *P < 0.05; **P < 0.01.

Close modal

To confirm the phenotype observed in the Cox6a2 KO mice, we further investigated the involvement of COX6A2 in islet cell apoptosis in vivo by delivering the AAV9-Cox6a2-KO (Cox6a2-AAV) virus or the control virus (vehicle) to diabetic GK rats. The administration of the AAV9-Cox6a2-KO virus resulted in decreased islet COX6A2 protein level (Supplementary Fig. 3A), which was accompanied by reduced cleaved caspase-3 and cleaved caspase-9 levels and TUNEL-positive and insulin-positive cells in GK rats (Fig. 4A–C and Supplementary Fig. 3BD). Accordingly, the insulin-positive cell area (Fig. 4D) and mass (Fig. 4E) in GK-Cox6a2-AAV rats were higher than those of GK-vehicle rats, respectively. Therefore, GK-Cox6a2-AAV rats maintained a lower blood glucose level (Fig. 4F), suggesting an improvement in diabetes.

Figure 4

Decreased COX6A2 suppresses β-cell apoptosis in GK rats. A: The levels of cleaved caspase-3 in islets from Wistar, GK-vehicle, and GK-Cox6a2-AAV rats. β-Actin was used as a loading control. Data are mean ± SEM (n = 6 rats per group). **P < 0.01. B: Immunofluorescence of TUNEL and insulin in pancreata from Wistar and GK rats administered an intraductal injection of vehicle or AAV9-Cox6a2-KO virus. Nuclei were stained with DAPI. Arrowheads indicate TUNEL-positive and insulin double-positive cells. Scale bar, 20 μm. Quantification of the TUNEL and insulin double-positive cell rate (C) and insulin-positive cell area (D) in B. Data are mean ± SEM (n = 6 rats per group). **P < 0.01. E: Statistical analysis of the β-cell mass in B. Data are mean ± SEM (n = 6 rats per group). **P < 0.01. F: Plasma glucose in Wistar, GK-vehicle, and GK-Cox6a2-AAV rats. Data are mean ± SEM (n = 6 rats per group). **P < 0.01 vs. GK-vehicle.

Figure 4

Decreased COX6A2 suppresses β-cell apoptosis in GK rats. A: The levels of cleaved caspase-3 in islets from Wistar, GK-vehicle, and GK-Cox6a2-AAV rats. β-Actin was used as a loading control. Data are mean ± SEM (n = 6 rats per group). **P < 0.01. B: Immunofluorescence of TUNEL and insulin in pancreata from Wistar and GK rats administered an intraductal injection of vehicle or AAV9-Cox6a2-KO virus. Nuclei were stained with DAPI. Arrowheads indicate TUNEL-positive and insulin double-positive cells. Scale bar, 20 μm. Quantification of the TUNEL and insulin double-positive cell rate (C) and insulin-positive cell area (D) in B. Data are mean ± SEM (n = 6 rats per group). **P < 0.01. E: Statistical analysis of the β-cell mass in B. Data are mean ± SEM (n = 6 rats per group). **P < 0.01. F: Plasma glucose in Wistar, GK-vehicle, and GK-Cox6a2-AAV rats. Data are mean ± SEM (n = 6 rats per group). **P < 0.01 vs. GK-vehicle.

Close modal

COX6A2 Promotes Cytochrome c Release From Mitochondria via Regulation of CypD

Given that COX6A2 is one of the subunits of mitochondrial complex IV (16), we examined the effect of COX6A2 overexpression on the ΔΨm. The vector control cells exhibited a higher ΔΨm, as indicated by stronger red and weaker green fluorescence. In contrast, COX6A2-overexpressing INS-1 832/13 cells displayed elevated green fluorescence, indicating a decrease in ΔΨm (Fig. 5A). Consequently, overexpression of COX6A2 promoted the release of cytochrome c from mitochondria to the cytoplasm (Fig. 5B) and hence activation of caspase-9 (Fig. 5C).

Figure 5

Increased COX6A2 promotes cytochrome c release from mitochondria via modulation of CypD. A: The ΔΨm of vector and COX6A2-overexpressing INS-1 832/13 cells were determined by JC-1 staining. Arrowheads indicated JC-1 monomers. Scale bar, 100 μm. B: The subcellular distribution of cytochrome c in vector and COX6A2-overexpressing INS-1 832/13 cells was determined by Western blotting. β-Actin and COX IV were used as internal control of cytosolic and mitochondrial protein, respectively. Data are mean ± SEM (n = 6) **P < 0.01. C: The protein levels of cleaved caspase-9 and caspase-9 in vector and COX6A2-overexpressing INS-1 832/13 cells. Data are mean ± SEM (n = 6). *P < 0.05. D: Colocalization of CypD and COX6A2 in INS-1 832/13 cells. Scale bar, 5 μm. E: Interaction between CypD and COX6A2 in vector and COX6A2-overexpressing INS-1 832/13 cells was immunoprecipitated (IP) with anti-CypD antibody, followed by Western blotting with the anti-COX6A2 antibody (n = 4). F: As in E, but coimmunoprecipitation was performed in INS-1 832/13 cells cultured in G5.5 or G30 medium for 72 h (n = 3). G: CypD protein levels in vector and COX6A2-overexpressing INS-1 832/13 cells were determined by Western blotting. β-Actin was used as a loading control. Data are mean ± SEM (n = 6). **P < 0.01. H: CypD protein expression in islets from Wistar, GK-vehicle, and GK-Cox6a2-AAV rats. Data are mean ± SEM (n = 6 rats per group). *P < 0.05, **P < 0.01. I: CypD protein level in islets from Wistar, GK-sham, and GK-RYGB rats. Data are mean ± SEM (n = 6 rats per group). *P < 0.05. J: The protein level of cleaved caspase-3 in vector and CypD-overexpressing INS-1 832/13 cells. β-Actin was used as a loading control. Data are mean ± SEM (n = 6). **P < 0.01. K: The subcellular distribution of cytochrome c in vector and COX6A2-overexpressing INS-1 832/13 cells treated with 5 μmol/L CsA for 24 h. L: Statistical analysis of the distribution of cytochrome c in K (n = 6). *P < 0.05, **P < 0.01. M: The cleaved caspase-3 level in vector and COX6A2-overexpressing INS-1 832/13 cells treated with 5 μmol/L CsA for 24 h. β-Actin was used as a loading control. Data are mean ± SEM (n = 6). *P < 0.05.

Figure 5

Increased COX6A2 promotes cytochrome c release from mitochondria via modulation of CypD. A: The ΔΨm of vector and COX6A2-overexpressing INS-1 832/13 cells were determined by JC-1 staining. Arrowheads indicated JC-1 monomers. Scale bar, 100 μm. B: The subcellular distribution of cytochrome c in vector and COX6A2-overexpressing INS-1 832/13 cells was determined by Western blotting. β-Actin and COX IV were used as internal control of cytosolic and mitochondrial protein, respectively. Data are mean ± SEM (n = 6) **P < 0.01. C: The protein levels of cleaved caspase-9 and caspase-9 in vector and COX6A2-overexpressing INS-1 832/13 cells. Data are mean ± SEM (n = 6). *P < 0.05. D: Colocalization of CypD and COX6A2 in INS-1 832/13 cells. Scale bar, 5 μm. E: Interaction between CypD and COX6A2 in vector and COX6A2-overexpressing INS-1 832/13 cells was immunoprecipitated (IP) with anti-CypD antibody, followed by Western blotting with the anti-COX6A2 antibody (n = 4). F: As in E, but coimmunoprecipitation was performed in INS-1 832/13 cells cultured in G5.5 or G30 medium for 72 h (n = 3). G: CypD protein levels in vector and COX6A2-overexpressing INS-1 832/13 cells were determined by Western blotting. β-Actin was used as a loading control. Data are mean ± SEM (n = 6). **P < 0.01. H: CypD protein expression in islets from Wistar, GK-vehicle, and GK-Cox6a2-AAV rats. Data are mean ± SEM (n = 6 rats per group). *P < 0.05, **P < 0.01. I: CypD protein level in islets from Wistar, GK-sham, and GK-RYGB rats. Data are mean ± SEM (n = 6 rats per group). *P < 0.05. J: The protein level of cleaved caspase-3 in vector and CypD-overexpressing INS-1 832/13 cells. β-Actin was used as a loading control. Data are mean ± SEM (n = 6). **P < 0.01. K: The subcellular distribution of cytochrome c in vector and COX6A2-overexpressing INS-1 832/13 cells treated with 5 μmol/L CsA for 24 h. L: Statistical analysis of the distribution of cytochrome c in K (n = 6). *P < 0.05, **P < 0.01. M: The cleaved caspase-3 level in vector and COX6A2-overexpressing INS-1 832/13 cells treated with 5 μmol/L CsA for 24 h. β-Actin was used as a loading control. Data are mean ± SEM (n = 6). *P < 0.05.

Close modal

CypD, a constitutive subunit of the mitochondrial permeability transition pore (mPTP), has been shown to mediate the release of cytochrome c from mitochondria (26,27). We hypothesized that CypD could participate in COX6A2-induced cytochrome c release. We, therefore, investigated the interaction between CypD and COX6A2. The immunofluorescence results showed that CypD colocalized with COX6A2 in INS-1 832/13 cells (Fig. 5D). Coimmunoprecipitation data confirmed the association between CypD and COX6A2, which was enhanced by COX6A2 overexpression (Fig. 5E). Similar observations were also made in high-glucose cultured INS-1 832/13 cells (Fig. 5F). We further examined the effect of COX6A2 on CypD expression. Notably, overexpression of COX6A2 resulted in elevated CypD protein expression (Fig. 5G), albeit it did not affect CypD mRNA level (Supplementary Fig. 4); consistently, CypD protein level was enhanced in GK-vehicle rat islets (Fig. 5H). However, the increased CypD protein level was markedly reduced in GK-Cox6a2-AAV rat islets (Fig. 5H). Similar observations were also made in GK-RYGB rat islets (Fig. 5I). These findings reveal that COX6A2 positively regulates CypD protein expression. Subsequently, the crucial role of CypD in cell apoptosis was confirmed by increased caspase-3 activation in CypD-overexpressing cells (Fig. 5J). Furthermore, inhibition of CypD by cyclosporin A (CsA) suppressed cytochrome c release from mitochondria to the cytoplasm induced by COX6A2 overexpression (Fig. 5K and L), thereby reducing the increased level of cleaved caspase-3 in COX6A2-overexpressing INS-1 832/13 cells (Fig. 5M), suggesting that CypD mediates the effects of COX6A2 on β-cell apoptosis.

GLP-1/PKA/ChREBP Axis Regulates COX6A2 Expression in β-Cells

To ascertain the mechanism behind the increased COX6A2 expression in islets from diabetic rats, we examined the impact of high glucose on COX6A2 expression. In line with the elevated expression of COX6A2 in GK islets, G30 led to increased COX6A2 mRNA (Fig. 6A) and protein (Fig. 6B) expression, respectively. ChREBP, which is known to be activated by high glucose, mediates gene expression changes induced by such conditions (12). We then explored the role of ChREBP in regulating COX6A2 expression. The results showed that transiently ectopic expression of ChREBP enhanced both mRNA (Fig. 6C) and protein (Fig. 6D) expression of COX6A2. On the contrary, ChREBP knockdown suppressed COX6A2 expression (Fig. 6E). To gain further understanding of the mechanism by which ChREBP regulates COX6A2 expression, we analyzed the DNA sequence in the Cox6a2 locus and identified a “CAGGTG” sequence as the putative ChREBP binding site (ChoRE) within the Cox6a2 promoter. We then constructed a luciferase reporter driven by the WT ChoRE or a mutant form “AGTTCT” (Fig. 6F) and assessed the luciferase activity. The data demonstrated that ChREBP increased reporter expression driven by the WT ChoRE but not by the mutant form (Fig. 6F), signifying that the ChoRE in the Cox6a2 promoter is essential for ChREBP-regulated COX6A2 expression. Moreover, the ChIP assay revealed that high glucose promoted ChREBP binding to the promoter of Cox6a2 (Fig. 6G) and increased the acetylation of histone H3 (ACH3), a marker associated with gene activation, at this promoter region (Fig. 6H). Considering that histone acetyltransferase p300 is a key enzyme facilitating ACH3 formation (28), we posited that p300 is involved in ACH3 production at the Cox6a2 promoter. Supporting this hypothesis, ChIP assays demonstrated an increased occupancy of p300 at the Cox6a2 promoter under high-glucose conditions (Fig. 6I). Notably, inhibition of p300 with its specific inhibitor C646 mitigated the aberrant expression of COX6A2 induced by high glucose (Fig. 6J), confirming that p300 mediates this process. These findings suggest that high glucose activates ChREBP, which then recruits p300 to enhance ACH3 at the Cox6a2 promoter, thereby upregulating COX6A2 expression in β-cells.

Figure 6

ChREBP regulates COX6A2 expression in β-cells. The mRNA (A) and protein (B) expression of COX6A2 in INS-1 832/13 cells cultured in G5.5 or G30 medium for 72 h. Data are mean ± SEM (n = 6–7). *P < 0.05, **P < 0.01. The mRNA (C) and protein (D) expression of COX6A2 in INS-1 832/13 cells transiently transfected with pcDNA3.1 (vector) or ChREBP plasmid. Data are mean ± SEM (n = 6). *P < 0.05, **P < 0.01.E: The COX6A2 protein expression in the scramble and ChREBP knockdown INS-1 832/13 cells. Data are mean ± SEM (n = 6). **P < 0.01. F: Human embryonic kidney 293 T cells were transfected with a luciferase reporter driven by WT or mutant ChREBP binding site in the presence of pcDNA3.1 or ChREBP plasmid, followed by determination of luciferase activity. Data are mean ± SEM (n = 8). **P < 0.01. G–I: INS-1 832/13 cells were cultured in G5.5 or G30 medium for 72 h. ChIP assays were performed to analyze the occupancy of ChREBP (G) and p300 (I), and the acetylation of histone H3 (H) at the promoter of Cox6a2. Data are mean ± SEM (n = 6). *P < 0.05, **P < 0.01. J: INS-1 832/13 cells were cultured in G5.5 or G30 medium in the presence of DMSO or 10 μmol/L p300 inhibitor C646 for 72 h, followed by Western blotting to examine COX6A2 protein expression. Data are mean ± SEM (n = 6). **P < 0.01. K: The protein level of phosphorylated (p)ChREBP and total ChREBP in islets from Wistar, GK-sham, and GK-RYGB rats. Data are mean ± SEM (n = 6 rats per group). *P < 0.05, **P < 0.01.

Figure 6

ChREBP regulates COX6A2 expression in β-cells. The mRNA (A) and protein (B) expression of COX6A2 in INS-1 832/13 cells cultured in G5.5 or G30 medium for 72 h. Data are mean ± SEM (n = 6–7). *P < 0.05, **P < 0.01. The mRNA (C) and protein (D) expression of COX6A2 in INS-1 832/13 cells transiently transfected with pcDNA3.1 (vector) or ChREBP plasmid. Data are mean ± SEM (n = 6). *P < 0.05, **P < 0.01.E: The COX6A2 protein expression in the scramble and ChREBP knockdown INS-1 832/13 cells. Data are mean ± SEM (n = 6). **P < 0.01. F: Human embryonic kidney 293 T cells were transfected with a luciferase reporter driven by WT or mutant ChREBP binding site in the presence of pcDNA3.1 or ChREBP plasmid, followed by determination of luciferase activity. Data are mean ± SEM (n = 8). **P < 0.01. G–I: INS-1 832/13 cells were cultured in G5.5 or G30 medium for 72 h. ChIP assays were performed to analyze the occupancy of ChREBP (G) and p300 (I), and the acetylation of histone H3 (H) at the promoter of Cox6a2. Data are mean ± SEM (n = 6). *P < 0.05, **P < 0.01. J: INS-1 832/13 cells were cultured in G5.5 or G30 medium in the presence of DMSO or 10 μmol/L p300 inhibitor C646 for 72 h, followed by Western blotting to examine COX6A2 protein expression. Data are mean ± SEM (n = 6). **P < 0.01. K: The protein level of phosphorylated (p)ChREBP and total ChREBP in islets from Wistar, GK-sham, and GK-RYGB rats. Data are mean ± SEM (n = 6 rats per group). *P < 0.05, **P < 0.01.

Close modal

Given the critical role of ChREBP in mediating COX6A2 expression, we evaluated the impact of RYGB on ChREBP phosphorylation. As shown in Fig. 6K, GK-sham rats displayed decreased ChREBP phosphorylation. However, RYGB led to enhanced ChREBP phosphorylation, suggesting an inhibitory effect of RYGB on ChREBP. As previous research has shown increased GLP-1 levels in patients after RYGB (8), and since PKA can phosphorylate ChREBP (14), we hypothesize that RYGB recovered ChREBP phosphorylation via an enhanced GLP-1 signaling pathway. To this end, we confirmed the increased GLP-1 levels in GK-RYGB rats (Supplementary Fig. 5A). Subsequently, we determined the stimulatory effect of GLP-1 signaling on ChREBP phosphorylation. As expected, the GLP-1R agonist exendin-4 or adenylate cyclase activator forskolin markedly promoted the phosphorylation of ChREBP (Fig. 7A and B), which was abolished by the specific PKA inhibitor Rp-8-Br-2’-O-MB-cAMPs (Rp-cAMP) (Fig. 7B). Given that the phosphorylation of ChREBP controls its nuclear translocation (14), we investigated the effect of exendin-4 on ChREBP subcellular distribution. Consistent with a previous study (29), high glucose promoted ChREBP translocation from the cytoplasm to the nucleus, which was inhibited by exendin-4 (Fig. 7C and D). Accordingly, exendin-4 abrogated high glucose–increased ACH3 at the Cox6a2 promoter (Fig. 7E), thereby diminishing high glucose–induced COX6A2 expression in a PKA-dependent manner (Fig. 7F and G and Supplementary Fig. 5B and C). Similar observations were also made in GK rats intraperitoneally injected with exendin-4 (Fig. 7H). As expected, exendin-4 treatment led to an increased insulin-positive cell area and alleviated hyperglycemia in GK rats (Supplementary Fig. 6AC).

Figure 7

GLP-1 signaling mediates RYGB-reduced COX6A2 expression in β-cells. A: INS-1 832/13 cells were treated with vehicle or 10 nmol/L exendin-4 for 72 h. ChREBP phosphorylation was examined by Western blotting. Data are mean ± SEM (n = 7). *P < 0.05. B: INS-1 832/13 cells were stimulated with 10 μmol/L forskolin or DMSO in the presence or absence of 30 μmol/L PKA inhibitor Rp-8-Br-2’-O-MB-cAMPs (Rp-cAMP) for 30 min, followed by Western blotting to determine ChREBP phosphorylation. Data are mean ± SEM (n = 6). **P < 0.01. C: INS-1 832/13 cells were cultured in G5.5 or G30 medium in the presence or absence of 10 nmol/L exendin-4 for 72 h. The cytosolic and nuclear localization of ChREBP was determined by Western blotting. β-Actin and histone H3 were used as the cytosolic and nuclear loading control, respectively. D: Quantification of ChREBP subcellular localization in C. Data are mean ± SEM (n = 6 independent experiments). *P < 0.05, **P < 0.01. E: INS-1 832/13 cells were cultured in the condition same as C. ChIP assay was performed to determine the acetylation of histone H3 at the Cox6a2 promoter. Data are mean ± SEM (n = 6). **P < 0.01. The mRNA (F) and protein (G) expression of COX6A2 in INS-1 832/13 cells cultured in the condition same as C. Data are mean ± SEM (n = 6). *P < 0.05, **P < 0.01. H: The COX6A2 protein level in pancreata from Wistar, GK-vehicle, and GK–exendin-4 rats. Data are mean ± SEM (n = 6). **P < 0.01. The protein levels of COX6A2 (I) and cleaved caspase-3 (J) in islets isolated from GK-RYGB-vehicle and GK-RYGB-exendin(9-39) rats. Data are mean ± SEM (n = 3 rats per group). *P < 0.05, **P < 0.01. K: Immunofluorescence of insulin and DAPI in pancreata of GK-RYGB-vehicle and GK-RYGB-exendin(9-39) rats. Scale bar, 200 μm. Data are mean ± SEM (n = 3 rats per group). *P < 0.05. L: Plasma insulin levels were determined in GK-RYGB-vehicle and GK-RYGB-exendin(9-39) rats following an intraperitoneal injection of glucose (1.5 g/kg body wt). Data are mean ± SEM (n = 3 rats per group). *P < 0.05, **P < 0.01. M: Plasma glucose in GK-RYGB-vehicle and GK-RYGB-exendin(9-39) rats. Data are mean ± SEM (n = 3). *P < 0.05, **P < 0.01.

Figure 7

GLP-1 signaling mediates RYGB-reduced COX6A2 expression in β-cells. A: INS-1 832/13 cells were treated with vehicle or 10 nmol/L exendin-4 for 72 h. ChREBP phosphorylation was examined by Western blotting. Data are mean ± SEM (n = 7). *P < 0.05. B: INS-1 832/13 cells were stimulated with 10 μmol/L forskolin or DMSO in the presence or absence of 30 μmol/L PKA inhibitor Rp-8-Br-2’-O-MB-cAMPs (Rp-cAMP) for 30 min, followed by Western blotting to determine ChREBP phosphorylation. Data are mean ± SEM (n = 6). **P < 0.01. C: INS-1 832/13 cells were cultured in G5.5 or G30 medium in the presence or absence of 10 nmol/L exendin-4 for 72 h. The cytosolic and nuclear localization of ChREBP was determined by Western blotting. β-Actin and histone H3 were used as the cytosolic and nuclear loading control, respectively. D: Quantification of ChREBP subcellular localization in C. Data are mean ± SEM (n = 6 independent experiments). *P < 0.05, **P < 0.01. E: INS-1 832/13 cells were cultured in the condition same as C. ChIP assay was performed to determine the acetylation of histone H3 at the Cox6a2 promoter. Data are mean ± SEM (n = 6). **P < 0.01. The mRNA (F) and protein (G) expression of COX6A2 in INS-1 832/13 cells cultured in the condition same as C. Data are mean ± SEM (n = 6). *P < 0.05, **P < 0.01. H: The COX6A2 protein level in pancreata from Wistar, GK-vehicle, and GK–exendin-4 rats. Data are mean ± SEM (n = 6). **P < 0.01. The protein levels of COX6A2 (I) and cleaved caspase-3 (J) in islets isolated from GK-RYGB-vehicle and GK-RYGB-exendin(9-39) rats. Data are mean ± SEM (n = 3 rats per group). *P < 0.05, **P < 0.01. K: Immunofluorescence of insulin and DAPI in pancreata of GK-RYGB-vehicle and GK-RYGB-exendin(9-39) rats. Scale bar, 200 μm. Data are mean ± SEM (n = 3 rats per group). *P < 0.05. L: Plasma insulin levels were determined in GK-RYGB-vehicle and GK-RYGB-exendin(9-39) rats following an intraperitoneal injection of glucose (1.5 g/kg body wt). Data are mean ± SEM (n = 3 rats per group). *P < 0.05, **P < 0.01. M: Plasma glucose in GK-RYGB-vehicle and GK-RYGB-exendin(9-39) rats. Data are mean ± SEM (n = 3). *P < 0.05, **P < 0.01.

Close modal

To further elucidate the role of the GLP-1 signaling pathway in the RYGB-decreased COX6A2 expression, GK-RYGB rats were intraperitoneally injected with the GLP-1R antagonist exendin(9-39) or saline. Notably, exendin(9-39) treatment resulted in increased expression of COX6A2 (Fig. 7I) and an elevated level of cleaved caspase-3 (Fig. 7J), alongside reduced insulin-positive cell area (Fig. 7K) within the islets of GK-RYGB rats compared with the saline-treated group. Consequently, rats treated with exendin(9-39) displayed lower plasma insulin levels (Fig. 7L) and higher blood glucose levels (Fig. 7M) compared with the saline control rats. These findings underscore the pivotal role of GLP-1 signaling in the RYGB-mediated inhibition of COX6A2 expression and β-cell apoptosis.

In this study, we demonstrate that RYGB reduces aberrant COX6A2 expression in islets from GK rats. The expression of COX6A2 is modulated by the GLP-1/PKA/ChREBP axis via a p300-mediated increase in ACH3 at the Cox6a2 promoter. For the first time, we show the proapoptotic effect of increased COX6A2 in β-cells through modulation of CypD (Fig. 8).

Figure 8

Schematic representation of the molecular mechanism of RYGB-inhibited COX6A2 expression and the role of COX6A2 in β-cell apoptosis. See text for details. ADCY, adenylate cyclase.

Figure 8

Schematic representation of the molecular mechanism of RYGB-inhibited COX6A2 expression and the role of COX6A2 in β-cell apoptosis. See text for details. ADCY, adenylate cyclase.

Close modal

The mitochondria-dependent cell apoptosis is characterized by decreased mitochondrial membrane potential, increased permeability of the mitochondrial membrane via opening mPTP, and release of cytochrome c under the stimulation of apoptotic signals. This then triggers apoptotic cascades, ultimately leading to apoptosis (30). Here, we demonstrated the crucial role of COX6A2 in promoting diabetic β-cell apoptosis, supported by the decreased cleaved caspase-3 and TUNEL immunofluorescence under suppressed or depleted COX6A2 levels. These observations may suggest that decreased β-cell apoptosis mediated by suppressed COX6A2 expression contributes to improved β-cell function, as indicated by increased β-cell mass, thus alleviating diabetic symptoms of hyperglycemia. Surprisingly, COX6A2 can interact with CypD. Moreover, CypD expression was positively correlated with COX6A2 expression. In the conditions where COX6A2 was overexpressed, there was higher CypD, higher cytochrome c release into the cytosol, and increased cleaved caspase-3 level. CypD plays a key role in mPTP function and promotes cell apoptosis via cytochrome c release, while inhibition suppresses cell apoptosis (26,31,32). The failure of COX6A2 overexpression to induce cytochrome c release and caspase-3 activation in the presence of CypD inhibitor CsA emphasized the essential role of CypD in COX6A2-mediated β-cell apoptosis. However, the molecular mechanism underlying COX6A2-induced CypD protein expression requires further exploration. Furthermore, given the constraints of in vivo models, further investigation is necessary to ascertain whether COX6A2 directly modulates β-cell apoptosis in vivo.

ChREBP is a glucose-response transcription factor that mediates glucose-induced gene expression. Exposure of β-cells to high glucose leads to an increase in ChREBP levels (33), and strikingly, increased ChREBP triggers β-cell apoptosis (12). However, the downstream target genes involved in ChREBP-mediated cell apoptosis remain largely unidentified. For the first time, we demonstrated that COX6A2 is one of the target genes regulated by ChREBP in β-cells. Three pieces of evidence support this view. First, activation or overexpression of ChREBP led to increased COX6A2 expression. Second, ChREBP knockdown suppressed COX6A2 expression. Third, there exists the ChoRE at the Cox6a2 promoter, and stimulation with high glucose resulted in increased ChREBP binding to the Cox6a2 promoter. Furthermore, our findings also revealed that activation of ChREBP by high glucose promoted the recruitment of the epigenetic regulator p300 and subsequently enhanced histone H3 acetylation at the Cox6a2 locus. This would lead to increased expression of COX6A2, given that acetylation of histones promotes gene transcription (28). Consistently, the p300 inhibitor C646 alleviated high glucose–induced COX6A2 expression, indicating that p300 is at least partially responsible for these effects of ChREBP. Therefore, the ChREBP/p300/COX6A2 axis may play a fundamental role in diabetic β-cell apoptosis.

Our data also demonstrated that RYGB downregulated COX6A2 expression in the islets of both diabetic GK rats and healthy SD rats. This downregulation would be a result of RYGB-induced inactivation of ChREBP through promoting ChREBP phosphorylation. RYGB has been proven to augment GLP-1 signaling (8–10). Correspondently, exendin-4 or forskolin phosphorylated ChREBP in a PKA-dependent manner. Moreover, the GLP-1R antagonist exendin(9-39) reversed the inhibitory effect of RYGB on COX6A2 expression. These data suggested that RYGB inhibits COX6A2 expression via modulation of the GLP-1/PKA/ChREBP signaling pathway. Therefore, RYGB likely reduces β-cell apoptosis by inhibiting COX6A2 expression.

In conclusion, our findings suggest that increased COX6A2 may facilitate β-cell apoptosis through the modulation of CypD-mediated cytochrome c release. The increase in COX6A2 expression could be a result of hyperglycemia-induced ChREBP overactivation in diabetes. RYGB is likely to reduce β-cell apoptosis by regulating the GLP-1/PKA/ChREBP/COX6A2 pathway. These findings not only uncover a novel way for regulating β-cell apoptosis but also highlight COX6A2 as a potential target for the treatment of T2D.

This article contains supplementary material online at https://doi.org/10.2337/figshare.27693066.

Funding. This work was funded by the National Natural Science Foundation of China (82070806, 82070845, 82370833, 82070978, 82072661), Natural Science Foundation of Guangdong Province, China (2023A1515010442), Shenzhen Science and Technology programs (JCYJ20210324094812033), and Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis (ZDSYS20190902092903237).

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. X.K. analyzed data and wrote the manuscript. X.K., D.Y., L.S., B.L., S.L., Y.T., and Y.Z. performed the experiments. X.K., X.S., Y.Y., and X.M. designed the experiments. X.K. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

1.
Méndez-Giménez
L
,
Becerril
S
,
Camões
SP
, et al
.
Role of aquaporin-7 in ghrelin- and GLP-1-induced improvement of pancreatic β-cell function after sleeve gastrectomy in obese rats
.
Int J Obes (Lond)
2017
;
41
:
1394
1402
2.
Otero
A
,
Becerril
S
,
Martín
M
, et al
.
Effect of guanylin peptides on pancreas steatosis and function in experimental diet-induced obesity and after bariatric surgery
.
Front Endocrinol (Lausanne)
2023
;
14
:
1185456
3.
Zhang
S
,
Guo
W
,
Wu
J
, et al
.
Increased β-cell mass in obese rats after gastric bypass: a potential mechanism for improving glycemic control
.
Med Sci Monit
2017
;
23
:
2151
2158
4.
Chai
F
,
Wang
Y
,
Zhou
Y
,
Liu
Y
,
Geng
D
,
Liu
J.
Adiponectin downregulates hyperglycemia and reduces pancreatic islet apoptosis after Roux-en-Y gastric bypass surgery
.
Obes Surg
2011
;
21
:
768
773
5.
Zhu
C
,
Xu
R
,
Li
Y
,
Andrade
M
,
Yin
DP.
Gastric bypass prevents diabetes in genetically modified mice and chemically induced diabetic mice
.
PLoS One
2021
;
16
:
e0258942
6.
Jones
B
,
Bloom
SR
,
Buenaventura
T
,
Tomas
A
,
Rutter
GA.
Control of insulin secretion by GLP-1
.
Peptides
2018
;
100
:
75
84
7.
Costes
S
,
Bertrand
G
,
Ravier
MA.
Mechanisms of beta-cell apoptosis in type 2 diabetes-prone situations and potential protection by GLP-1-based therapies
.
Int J Mol Sci
2021
;
22
:
5303
8.
Laferrère
B
,
Heshka
S
,
Wang
K
, et al
.
Incretin levels and effect are markedly enhanced 1 month after Roux-en-Y gastric bypass surgery in obese patients with type 2 diabetes
.
Diabetes Care
2007
;
30
:
1709
1716
9.
Lindqvist
A
,
Spégel
P
,
Ekelund
M
, et al
.
Gastric bypass improves β-cell function and increases β-cell mass in a porcine model
.
Diabetes
2014
;
63
:
1665
1671
10.
Kong
X
,
Feng
L
,
Yan
D
,
Li
B
,
Yang
Y
,
Ma
X.
FXR-mediated epigenetic regulation of GLP-1R expression contributes to enhanced incretin effect in diabetes after RYGB
.
J Cell Mol Med
2024
;
28
:
e16339
11.
Zhang
P
,
Kumar
A
,
Katz
LS
, et al
.
Induction of the ChREBPβ isoform is essential for glucose-stimulated β-cell proliferation
.
Diabetes
2015
;
64
:
4158
4170
12.
Poungvarin
N
,
Lee
JK
,
Yechoor
VK
, et al
.
Carbohydrate response element-binding protein (ChREBP) plays a pivotal role in beta cell glucotoxicity
.
Diabetologia
2012
;
55
:
1783
1796
13.
Cha-Molstad
H
,
Saxena
G
,
Chen
J
,
Shalev
A.
Glucose-stimulated expression of Txnip is mediated by carbohydrate response element-binding protein, p300, and histone H4 acetylation in pancreatic beta cells
.
J Biol Chem
2009
;
284
:
16898
16905
14.
Abdul-Wahed
A
,
Guilmeau
S
,
Postic
C.
Sweet sixteenth for ChREBP: established roles and future goals
.
Cell Metab
2017
;
26
:
324
341
15.
Nagai
Y
,
Matsuoka
T-A
,
Shimo
N
, et al
.
Glucotoxicity-induced suppression of Cox6a2 expression provokes β-cell dysfunction via augmented ROS production
.
Biochem Biophys Res Commun
2021
;
556
:
134
141
16.
Quintens
R
,
Singh
S
,
Lemaire
K
, et al
.
Mice deficient in the respiratory chain gene Cox6a2 are protected against high-fat diet-induced obesity and insulin resistance
.
PLoS One
2013
;
8
:
e56719
17.
Sanz-Morello
B
,
Pfisterer
U
,
Winther Hansen
N
, et al
.
Complex IV subunit isoform COX6A2 protects fast-spiking interneurons from oxidative stress and supports their function
.
Embo J
2020
;
39
:
e105759
18.
Li
X
,
Jiang
L
,
Yang
M
,
Wu
Y-w
,
Sun
S-x
,
Sun
J-z.
Expression of CTRP3, a novel adipokine, in rats at different pathogenic stages of type 2 diabetes mellitus and the impacts of GLP-1 receptor agonist on it
.
J Diabetes Res
2014
;
2014
:
398518
19.
Lin
P-I
,
Tai
Y-T
,
Chan
WP
,
Lin
Y-L
,
Liao
M-H
,
Chen
R-M.
Estrogen/ERα signaling axis participates in osteoblast maturation via upregulating chromosomal and mitochondrial complex gene expressions
.
Oncotarget
2018
;
9
:
1169
1186
20.
Yorek
MA.
Alternatives to the streptozotocin-diabetic rodent
.
Int Rev Neurobiol
2016
;
127
:
89
112
21.
Kong
X
,
Tu
Y
,
Li
B
, et al
.
Roux-en-Y gastric bypass enhances insulin secretion in type 2 diabetes via FXR-mediated TRPA1 expression
.
Mol Metab
2019
;
29
:
1
11
22.
Oertel
M
,
Ziegler
CG
,
Kohlhaas
M
, et al
.
GLP-1 and PYY for the treatment of obesity: a pilot study on the use of agonists and antagonists in diet-induced rats
.
Endocr Connect
2024
;
13
:
e230398
23.
Luo
G
,
Kong
X
,
Lu
L
,
Xu
X
,
Wang
H
,
Ma
X.
Glucagon-like peptide 1 potentiates glucotoxicity-diminished insulin secretion via stimulation of cAMP-PKA signaling in INS-1E cells and mouse islets
.
Int J Biochem Cell Biol
2013
;
45
:
483
490
24.
Mo
H
,
Liu
Y
,
Zhang
M
, et al
.
The role of pancreatic fatty acid synthesis in islet morphology and function after caloric restriction or Roux-en-Y gastric bypass surgery in mice
.
Genes (Basel)
2022
;
14
:
5
25.
Kong
X
,
Zhang
L
,
Hua
X
,
Ma
X.
Squamosamide derivative FLZ protects pancreatic β-cells from glucotoxicity by stimulating Akt-FOXO1 pathway
.
J Diabetes Res
2015
;
2015
:
803986
26.
Kumarswamy
R
,
Chandna
S.
Putative partners in Bax mediated cytochrome-c release: ANT, CypD, VDAC or none of them?
Mitochondrion
2009
;
9
:
1
8
27.
Yang
H
,
Li
R
,
Zhang
L
, et al
.
P53-cyclophilin D mediates renal tubular cell apoptosis in ischemia-reperfusion-induced acute kidney injury
.
Am J Physiol Renal Physiol
2019
;
317
:
F1311
F1317
28.
Haery
L
,
Thompson
RC
,
Gilmore
TD.
Histone acetyltransferases and histone deacetylases in B- and T-cell development, physiology and malignancy
.
Genes Cancer
2015
;
6
:
184
213
29.
Denechaud
P-D
,
Bossard
P
,
Lobaccaro
J-MA
, et al
.
ChREBP, but not LXRs, is required for the induction of glucose-regulated genes in mouse liver
.
J Clin Invest
2008
;
118
:
956
964
30.
Sinha
K
,
Das
J
,
Pal
PB
,
Sil
PC.
Oxidative stress: the mitochondria-dependent and mitochondria-independent pathways of apoptosis
.
Arch Toxicol
2013
;
87
:
1157
1180
31.
Yan
S
,
Du
F
,
Wu
L
, et al
.
F1F0 ATP synthase-cyclophilin D interaction contributes to diabetes-induced synaptic dysfunction and cognitive decline
.
Diabetes
2016
;
65
:
3482
3494
32.
Fujimoto
K
,
Chen
Y
,
Polonsky
KS
,
Dorn
GW.
Targeting cyclophilin D and the mitochondrial permeability transition enhances beta-cell survival and prevents diabetes in Pdx1 deficiency
.
Proc Natl Acad Sci U S A
2010
;
107
:
10214
10219
33.
Chau
GC
,
Im
DU
,
Kang
TM
, et al
.
mTOR controls ChREBP transcriptional activity and pancreatic β cell survival under diabetic stress
.
J Cell Biol
2017
;
216
:
2091
2105
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/journals/pages/license.